JP2004528314A - Rho kinase inhibitor - Google Patents

Rho kinase inhibitor Download PDF

Info

Publication number
JP2004528314A
JP2004528314A JP2002576235A JP2002576235A JP2004528314A JP 2004528314 A JP2004528314 A JP 2004528314A JP 2002576235 A JP2002576235 A JP 2002576235A JP 2002576235 A JP2002576235 A JP 2002576235A JP 2004528314 A JP2004528314 A JP 2004528314A
Authority
JP
Japan
Prior art keywords
compound
formula
rho kinase
acid
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
JP2002576235A
Other languages
Japanese (ja)
Other versions
JP4320705B2 (en
Inventor
ナガラスナム,ダナパラン
ワン,チュングアン
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer Corp
Original Assignee
Bayer Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bayer Corp filed Critical Bayer Corp
Publication of JP2004528314A publication Critical patent/JP2004528314A/en
Application granted granted Critical
Publication of JP4320705B2 publication Critical patent/JP4320705B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Abstract

化合物およひその誘導体、それらの合成、およびRhoキナーゼ阻害剤としてそれらの使用が開示される。本発明のこれらの化合物は腫瘍発育の阻止、勃起不全の処置、およびRhoキナーゼによって仲介される他の適応症、例えば冠心疾患の処置のために有用である。Compounds and their derivatives, their synthesis, and their use as Rho kinase inhibitors are disclosed. These compounds of the present invention are useful for inhibiting tumor growth, treating erectile dysfunction, and other indications mediated by Rho kinase, such as coronary heart disease.

Description

【技術分野】
【0001】
本出願は2001年3月23日に出願された米国仮出願No.60/277,974および2001年8月29日に出願された米国特許出願No.60/315,338の出願日の利益を主張する。
【0002】
本発明は化合物、その誘導体、それらの合成、およびRhoキナーゼ阻害剤としてそれらの使用に関する。本発明のこれらの化合物は腫瘍発育阻止、勃起不全の処置、およびRhoキナーゼによって仲介される他の適応症、例えば冠心疾患の処置に有用である。
【背景技術】
【0003】
高血圧、勃起不全、冠大脳循環障害、神経退化障害およびがんを含む多数のヒトおよび動物病の病理学は、アクチン細胞骨格の変化に直接結びつけることができる。これらの病気は重大な満たされない医学的必要性を提供する。アクチン細胞骨格はすべての真核細胞に見られるアクチンフィラメントの網とアクチン結合タンパクから構成されている。平滑筋細胞においては、アクチン細胞骨格の集合および離散が平滑筋収縮および弛緩に責任ある一次的原動力である。非平滑筋細胞においては、アクチン細胞骨格の動的再配置が細胞形態、細胞移動性、アクチンストレス線維生成、細胞接着、および軸索収縮、ファゴサイト−シスもしくは細胞分裂のような特化された細胞機能の調節に責任がある(Van Aelst,et al.Genes Dev 1997,11,2295)。
【0004】
アクチン細胞骨格はRasスーパーファミリーのサブセットであるタンパクのファミリーによってコントロールされる。このサブセットは現在RhoAないしGとRhoG(集合的にRhoと呼ばれる)、Rac1および2,Cdc42HsおよびG25KおよびTC10イソフォームよりなる(Mackay,et al.J Bio Chem 1998,273,20685)。これら3種のタンパクは固有のGTPアーゼ活性を持つGTP(グアニンヌクレオチドトリフォスフェート)結合タンパクである。それらは分子スイッチとして作用し、そして結合したGDP(グアニンヌクレオチドジフォスフェート)とそして活性GTP結合状態の間をサイクルにする。生化学的および遺伝子操作を使用して、各ファミリーメンバーに機能を割当てることが可能になった。活性化した時Rhoタンパクはアクチンストレス線維とアクチンフィラメントの厚い束の生成、そして焦点の接着コンプレックスにおいてインテグリンのクラスター化を制御する。活性化した時Racタンパクは細胞表面上の膜波打ちの生成を制御し、そしてCdc42は糸状足生成を制御する。合同してこれらタンパクファミリーは細胞運動、軸索誘導、細胞分裂、そして細胞形態、形状および極性の変化を含む、キー細胞機能の制御において重要な役割を果す。
【0005】
細胞のタイプおよび活性化レセプターに応じて、Rhoタンパクは様々の生物学的応答を制御することができる。平滑筋細胞においては、Rhoタンパクは平滑筋収縮の間カルシウム感作に責任がある。非平滑筋細胞においては、Rho GTPアーゼはリソフォスファチジン酸(LPA)、トロンビンおよびトロンボキサンA2のようなアゴニストに対する細胞応答に対して責任がある(Fukuda,et al.Trends Pharol Sci 2001,22,32)。アゴニスト応答は、他のレセプターも含まれ得るが、ヘテロトリマーGタンパク、Gアルファ12またはGアルファ13を介して連結される(Goetzl,et al.Cancer Res 1999,59,4732;Buhl,et al.J Biol Chem 1995,270,24681)。活性化した時Rho GTPアーゼはPIP5−キナーゼ、ローセキン、ローフィリン、PKNおよびRhoキナーゼイソフォームROCK−1/ROKベーターおよびROCK−1/ROKアルファを含む多数の下流エファクターを活性化する(Mackay et al.J Biol Chem 1998,273,20685;Aspenstrom,Curr Opin Cell Biol 1999,11,95;Amano,et al.Exp Cell Res 2000,261,44)。
【0006】
Rhoキナーゼはウシ脳から単離されたRhoA相互作用タンパクとして同定された(Matsui,et al.Embo J 1996,15,2208)。それはタンパクキナーゼの筋ジストロフィーファミリーのメンバーであり、そしてアミノ末端においてセリン/スレオニンドメインと、中央領域においてコイル巻きドメインと、そしてカルボキシ末端においてRho相互作用ドメインを含んでいる(Amano,et al.Exp Cell Res 2000,261,44)。そのキナーゼ活性はGTP結合RhoAへ結合する時増強され、そして細胞へ導入される時それは活性化RhoAの多数の活性を再現する。平滑筋細胞においては、Rhoキナーゼはカルシウム感作および平滑筋収縮を仲介し、Rhoキナーゼの阻害は筋収縮によって誘発された5−HTおよびフェニレフリンアゴニストをブロックする。非平滑筋細胞へ導入された時、Rhoキナーゼはストレス線維生成を誘発し、そしてRhoAによって仲介される細胞形質転換のために必要である(Sahai,et al.Curr Biol 1999,9,136)。Rhoキナーゼは、フォスフォリル化によりミオシン軽鎖(Somlyo,et al.J Physio(Lond)2000,522 pt2,177)、ミオシン軽鎖フォファターゼ結合サブユニット(Fukata,et al.J Cell Biol 1998,141,409)、およびLIM−キナーゼ2(Sumi,et al.J Bio Chem 2001,276,670)を含む多数の下流タンパクを調節する。
【0007】
動物モデルにおいてRhoキナーゼの阻害はヒト病気のためのRhoキナーゼ阻害剤の多数の利益を証明した。(+)−トランス−4−(1−アミノエチル)−(ピリジン−4−イルアミノカルボニル)シクロヘキサンジ塩酸塩−水和物(WO00/078351,WO00/057913)、および置換イソキノリンスルホニル化合物(EP00187371)を動物モデルにおいて活性を有するRhoキナーゼ阻害剤としてクレームしているいくつかの特許が見られる。これらは高血圧(Vehata,et al.Nature 1997,389,990)、動脈硬化(Retzer,et al.FEBS Lett 2000,466,70)、再狭窄(Eto,et al.Am J Physiol Heart Circ Physiol 2000,H1744;Negoro,et al.Biochem Biophys Res Commun 1999,261,211)、脳虚血症(Uehata et al.Nature 1997,389,990);Seasholtz,et al.Circ Res 1999,84,1186;Hitomi,et al.Life Sci 2000,67,1929;Yamamoto,et al.J Cardiovasc Phamacol 2000,35,203),脳血管痙れん(Sato,et al.Circ Res 2000,87,195;Kim,et al.Neurosurgery 2000,46,440),勃起不全(Chitaley,et al.Nat Med 2001,7,119),神経退化および脊髄障害のような中枢神経系障害(Hara,et al.J Neurosurg 2000,93,94;Toshima,et al.Stroke 2000,31,2245),Rhoキナーゼの阻害が腫瘍細胞生育および転移を阻害することを示した新形成(Itoh,et al.Nat Med 1999,5,221;Somlye,et al.Biochem Biophys Res Commun 2000,269,652),脈管形成(Uchida,et al.Biochem Biophys Res Commun 2000,269,633;Gingras,et al.Biochem J 2000,348 Pt 2,273),血小板凝集(Klages,et al.J Cell Biol 1999,144,745;Retzer,et al.Cell Signal 2000,12,645)および白血球凝集(Kawaguchi,et al.Eur J Phamacol 2000,403,203;Sanchez−Madrid,et al.Embo J 1999,18,501)のような動脈止血障害、喘息(Setoguchi,et al.Br J Phamacol 2001,132,111;Nakahara,et al.Eur J Phamacol 2000,389,103),眼内圧調節(Honjo,et al.Invest Ophthalmol Vis Sci 2001,42,137),および骨吸収(Chellaiah,et al.J Biol Chem 2000,275,11993;Zhang,et al.J Cell Sci 1995,108,2285)のような循環器病のモデルを含んでいる。
【0008】
患者においてRhoキナーゼの阻害はクモ膜下出血後の脳血管痙れんおよび虚血を制御するための利益を有する(Pharma Japan 1995,1470,16)。
【発明の開示】
【0009】
本発明において提供される化合物およびそれらの誘導体はRhoキナーゼ阻害剤として有用であり、そのため高血圧、動脈硬化、再狭窄、脳虚血病、脳血管痙れん、神経退化、骨髄損傷、乳房、前立腺、大腸、卵巣、脳および肺のがんおよびそれらの転移、止血障害、喘息、緑内障および骨粗しょう症の処理に有用性を有する。
【0010】
加えて、本発明の化合物は勃起不全、すなわちRhoキナーゼによって仲介される勃起不全を処置するために有用である。勃起不全は性交に適切な勃起を得ることまたは持続することの不可能として定義することができる。WO94/28902,U.S.P.6,103,765およびU.S.P.6,124,461参照。
【0011】
本発明は以下の式の化合物を提供する。
【0012】
【化15】

Figure 2004528314
【0013】
【化16】
Figure 2004528314
【0014】
【化17】
Figure 2004528314
【0015】
【化18】
Figure 2004528314
【0016】
【化19】
Figure 2004528314
【0017】
【化20】
Figure 2004528314
【0018】
本発明は式I−VIの薬学的に許容し得る塩にも向けられる。好適な薬学的に許容し得る塩は当業者には良く知られており、そして塩酸、臭化水素酸、硫酸、リン酸、メタンスルホン酸、スルホン酸、酢酸、トリフルオロ酢酸、リンゴ酸、酒石酸、クエン酸、乳酸、シュウ酸、コハク酸、フマル酸、マレイン酸、安息香酸、サリチル酸、フェニル酢酸、およびマンデル酸のような無機酸および有機酸の塩を含む。加えて、薬学的に許容し得る塩は、アルカリカチオン(例えばLi+,Na+またはK+)、アルカリ土類カチオン(例えばMg++,Ca++,またはBa++)、アンモニウムカチオンを含んでいる塩のような無機塩基の酸塩、それにトリエチルアミン、N,N−ジエチルアミン、N,N−ジシクロヘキシルアミン、ピリジン、N,N−ジメチルアミノピリジン(DMAP)、1,4−ジアザビシクロ〔2.2.2〕オクタン(DABCO)、1,5−ジアザビシクロ〔4.3.0〕ノネンー5(DBN)および1,8−ジアザビシクロ〔5.4.0〕ウンデセン−7(BBU)のプロトン化またはペルアルキル化から発生するような脂肪族および芳香族置換アンモニウムおよび4級アンモニウムカチオンを含んでいる有機塩基の酸塩を含む。
【0019】
式I−VIの化合物の多数は不斉炭素原子を有し、そしてそれ故ラセミ形および光学活性形で存在する。エナンチオマーおよびジアステレオマー混合物の分離方法は当業者には良く知られている。本発明はRhoキナーゼ阻害活性を有する式I−VIの化合物の単離されたラセミ形または光学活性形のどれも含む。
【0020】
本発明は、式I−VIの化合物と、生理学的に許容し得る担体を含んでいる薬剤組成物も含んでいる。
【0021】
さらに本発明は、式I−VIの化合物、または式I−VIの化合物を含む薬剤組成物を投与することにより、Rhoキナーゼによって仲介される適応症を処置することを含む。このように、本発明は高血圧、動脈硬化、再狭窄、脳虚血症または痙れんのような循環器病、神経退化および脊髄傷害のような中枢神経系障害、勃起不全例えばPDE−5に対して満足な応答を持たない患者、およびRhoキナーゼによって仲介されるがん(例えば腫瘍生育)に対し、式Iの化合物の有効量を例えばそれを必要とする宿主へ投与することによって処置することを含む。Rhoキナーゼによって仲介されるがんおよび腫瘍は、乳房、大腸、前立腺、卵巣、脳および肺のがんと、それらの転移を含む。
【0022】
化合物は、投与単位製剤において経口、局所、非経口、吸入またはスプレー、経膣、経腸または舌下的に投与することができる。注射によって投与とは、静脈内、動脈内、筋肉内、皮下および非経口注射と、それに注入技術の使用も含む。皮膚投与は局所的または経皮投与を含み得る。1以上の化合物が1種以上の非毒性の薬学的に許容し得る担体および、もし望むならば、他の活性成分と共に存在し得る。
【0023】
経口使用を意図する組成物は、薬剤組成物の製造のための任意の適切な既知方法に従って製造することができる。そのような組成物は服用し得る製剤を提供するため、希釈剤、甘味剤、矯味剤、着色剤および保存剤からなる群から選ばれた1以上の剤を含有することができる。錠剤は錠剤の製造に適した非毒性の薬学的に許容し得る補助剤と混合された活性成分を含む。これら補助剤は、例えば、炭酸カルシウム、炭酸ナトリウム、乳糖、リン酸カルシウムまたはリン酸ナトリウムのような不活性希釈剤、造粒剤および崩壊剤、例えばコーンスターチまたはアルギン酸、および結合剤、例えばステアリン酸マグネシウム、ステアリン酸またはタルクである。錠剤は未被覆か、または消化管内において崩壊および吸収を遅らせ、それによって長時間に亘って被覆することができる。例えば、グリセリルモノステアレートまたはグリセリルジステアレートのような時間遅延材料を使用することができる。これら化合物は固形の速放性形にも調製することができる。
【0024】
経口使用のための製剤は硬ゼラチンカプセルとしても提供することができ、その場合は活性成分は不活性の固体希釈剤、例えば炭酸カルシウム、リン酸カルシウムまたはカオリンと混合され、または活性成分が水または油性媒体、例えばピーナッツ油、液状パラフィンもしくはオリーブ油と混合される軟ゼラチンカプセルとして提供することもできる。
【0025】
水性懸濁液の製造のために適した補助剤との混合物に活性成分を含んでいる水性懸濁液も使用することができる。そのような補助剤は懸濁剤、例えばカルボキシメチルセルロースナトリウム、メチルセルロース、ヒドロキシプロピルメチルセルロース、アルギン酸ナトリウム、ポリビニルピロリドン、トラガントガムおよびアラビアガムであり、分散剤もしくは湿潤剤は天然フォスファチド例えばレシチン、または脂肪酸のアルキレンオキサイド縮合物例えばポリオキシエチレンステアレート、または長鎖脂肪族アルコールとのアルキレンオキサイド縮合物例えばヘプタデカエチレンオキシセタノール、またはポリオキシエチレンソルビトールモノオレエートのような脂肪酸とヘキシトールから誘導された部分エステルのアルキレンオキシド縮合物、または脂肪酸とヘキシトール無水物から誘導された部分エステルのアルキレンオキシド縮合物例えばポリオキシエチレンソルビタンモノオレエートであることができる。水性懸濁液はまたは1種以上の保存剤例えばp−ヒドロキシ安息香酸エチルもしくはn−プロプルエステル、1種以上の着色剤、1種以上の矯味剤、およびショ糖もしくはサッカリンのような1種以上の甘味剤を含むことができる。
【0026】
水の添加によって水性懸濁液の調製に適した分散性粉末もしくは顆粒は、分散剤もしくは湿潤剤、懸濁剤および1種以上の保存剤との混合物中に活性成分を提供する。好適な分散剤もしくは湿潤剤および懸濁剤は上で述べたものによって例示されている。追加の補助剤例えば甘味剤、矯味剤および着色剤も存在してよい。
【0027】
化合物は非水性液状製剤の形、例えば活性成分を植物油例えばアラキス油、オリーブ油、ゴマ油もしくはピーナッツ油中に、または液状パラフィンのような鉱物油中に懸濁することにより製剤化することができる油性懸濁液でもよい。油性懸濁液は増粘剤例えば蜜ロウ、硬パラフィンまたはセチルアルコールを含むことができる。上で述べたような甘味剤および矯味剤を服用容易な経口製剤を提供するために添加することができる。これらの組成物はアスコルビン酸のような抗酸化剤の添加によって保存することができる。
【0028】
本発明の化合物は当業者に既知の方法 (例えば、Chien;“Transdermal Controlled Systemic Medications”;Marcel Dekker,Inc,;1987.Lipp et al.WO94/04157,1994年3月3日を見よ。)を使用して経皮的に投与することもできる。例えば、任意に浸透促進剤を含有する適当な揮発性溶媒中の式I−VIの化合物の溶液もしくは懸濁液をマトリックス材料および殺バクテリア剤のような当業者に既知の追加の添加剤と組合わせることができる。滅菌後、生成した混合物は既知の操作に従って投与形に製剤化することができる。加えて、乳化剤および水との処理において、式I−VIの化合物の溶液もしくは懸濁液をローションもしくはザルベに処方することができる。
【0029】
経皮デリバリシステムへ加工するために適した溶媒は当業者に既知であり、そしてエタノールもしくはプロピルアルコールのような低級アルコール、アセトンのような低級ケトン、酢酸エチルのような低級カルボン酸エステル、テトラヒドロフランのような極性エーテル、ヘキサン、シクロヘキサンもしくはベンゼンのような低級炭化水素、またはジクロロメタン、クロロホルム、トリクロロトリフロロエタンもしくはトリクロロフロロエタンのようなハロゲン化炭化水素を含む。好適な溶媒は低級アルコール、低級ケトン、低級カルボン酸エステル、極性エーテル、低級炭化水素およびハロゲン化炭化水素から選ばれた1種以上の材料の混合物を含むことができる。
【0030】
経皮デリバリシステムのために適した浸透促進剤は当業者に既知であり、そして例えばエタノール、プロピレングリコールもしくはベンジルアルコールのようなモノヒドロキシまたはポリヒドロキシアルコール、ラウリルアルコールもしくはセチルアルコールのような飽和もしくは不飽和C8−C18脂肪アルコール、ステアリン酸のような飽和もしくは不飽和C8−C18脂肪酸、酢酸、カプロン酸、ラウリン酸、ミリスチン酸、ステアリン酸もしくはパルミチン酸、メチル、エチル、プロピル、イソプロピル、n−ブチル、sec−ブチル、イソブチル、t−ブチルもしくはモノグリセリンエステルのような24までの炭素原子を有する飽和もしくは不飽和脂肪酸エステル、またはジイソプロピルアジペート、ジイソブチルアジペート、ジイソプロピルセバケート、ジイソプロピルマレエートまたはジイソプロピルフマレートのような24までの炭素原子を有する飽和もしくは不飽和脂肪酸のジエステルを含む。追加の浸透促進剤はレシチンもしくはセファリンのようなフォスファチジル誘導体、テルペン、アミド、ケトン、尿素およびその誘導体、およびジメチルイソソルビッドおよびジエチレングリコールモノエチルエーテルのようなエーテルを含む。好適な浸透促進剤はモノヒドロキシもしくはポリヒドロキシアルコール、飽和もしくは不飽和C8−C18脂肪アルコール、飽和もしくは不飽和C8−C18脂肪酸、24までの炭素原子を有する飽和もしくは不飽和脂肪酸エステル、合計24までの炭素を有する飽和もしくは不飽和ジカルボン酸のジエステル、フォスファチジル誘導体、テルペン、アミド、ケトン、尿素およびその誘導体およびエーテルから選ばれた1種以上の材料の混合物を含むことができる。
【0031】
経皮デリバリシステムのための好適な結合材料は当業者に既知であり、そしてポリアクリレート、シリコーン、ポリウレタン、ブロックコポリマー、スチレンブタジエンコポリマー、天然および合成ゴムを含む。セルロースエーテル、誘導体化ポリエチレンおよびシリケートもマトリックス成分として使用することができる。粘性樹脂もしくはオイルのような追加の添加剤をマトリックスの粘度を上げるために添加することができる。
【0032】
本発明の薬剤組成物はO/Wエマルジョンの形でもよい。油相は例えばアラキス油もしくはオリーブ油のような植物油、または鉱油例えば液状パラフィン、またはこれらの混合物でよい。好適な乳化剤は天然ガム例えばアラビアガムもしくはトラガントガム、天然フォスファチジド例えば大豆レシチン、脂肪酸とヘキシトール無水物から誘導されたエステルもしくは部分エステル例えばソルビタンモノオレエート、および前記部分エステルとエチレンオキシドとの縮合物例えばポリオキシエチレンソルビタンモノオレエートであることができる。エマルジョンは甘味剤および矯味剤を含むことができる。
【0033】
シロップおよびエリキサーは甘味剤例えばグリセロール、プロピレングリコール、ソルビトールもしくはショ糖と共に処方することができる。そのような処方は刺激緩和剤、保存剤、矯味剤および着色剤を含むことができる。
【0034】
化合物は薬物投与のため経直腸もしくは経膣坐剤の形で投与することもできる。これら組成物は常温では固体であるが、しかし直腸内温度もしくは膣内温度では液体であり、そして薬物を放出するように直腸もしくは膣内で溶融する適当な非刺激性補助剤と薬物を混合することによって調製することができる。そのような材料はココアバターおよびポリエチレングリコールを含む。
【0035】
さらに勃起不全の処置のためには、本薬剤組成物は陰核海綿体中への注射もしくは経尿道投与により、または尿道へ局所的に適用することによりペニスへ投与するのに適した任意の形を取ることができる。陰核海綿体への注射の場合、薬剤組成物は好適には食塩水溶液である。好ましくは薬剤組成物は経尿道投与に適した形であり、そしてこの場合組成物は典型的には溶液、軟膏もしくは坐剤の形である。典型的には薬剤組成物は性交開始前1ないし50分、好ましくは10ないし20分前に投与される。
【0036】
式Iの化合物のためのここに開示した使用のすべての療法において、毎日の経口投与療法は総体重kgあたり好ましくは0.01ないし200mgであろう。
【0037】
静脈内、筋肉内、皮下および非経口注射を含む注射による、そして注入技術の使用による投与のための1日投与量は好ましくは総体重kgあたり0.01ないし200mgであろう。毎日の経膣療法は好ましくは総体重kgあたり0.01ないし200mgであろう。毎日の局所投与療法は好ましくは1日1ないし4回投与される総体重kgあたり0.01ないし200mgであろう。経皮濃度は好ましくは0.1ないし200mg/kgの1日投与量を維持するのに要する濃度であろう。毎日の吸入投与療法は好ましくは総体重kgあたり0.01ないし10mgであろう。
【0038】
当業者には、特定の投与方法は治療薬を投与する時日常的に考慮される種々の要因に依存することが認められるであろう。しかしながら与えられた患者のための特定投与量レベルは、使用される特定化合物の活性、患者の年令、患者の体重、患者の一般的健康状態、患者の性、患者の食事、投与時間、投与ルート、排泄速度、薬物組合せ、および治療を受けている症状の重篤度を含む種々の要因に依存することも理解されるであろう。さらに当業者には、最適な処置コース、すなわち処置モードおよび限られた日数にわたって投与される式Iの化合物もしくは薬学的に許容されるその塩の1日あたりの投与回数は慣用の処置テストを使用して確かめることができることが認められるであろう。
【0039】
本化合物および組成物はRhoのキナーゼ阻害活性を示し、そのため上で挙げた適応症、例えばRhoキナーゼによって仲介される適応症の処置に有用である。Rhoキナーゼによって仲介される適応症とは、その進行が少なくとも一部Rho経路を経由して進行する病気または疾状を意味する。
【0040】
Rhoキナーゼ阻害活性、例えばROCK−1阻害は以下のように評価することができる。
【0041】
ヒトROCKのキナーゼドメインであるアミノ酸27−530がSf9こん虫細胞からグルタチオンS−トランフフェラーゼ融合タンパクとして単離される。このタンパクがグルタチオンセファロース4B(Pharmacia Biotech,NJ)アフィニティ精製により部分的に精製される。反応は50mM N−(2−ヒドロキシエチル)ピペラジン−N’−(2−エタンスルホン酸)pH7.5、5μM MgCl2,1mMジチオスレリトール、6μM ATP, 0.2μCi〔33P〕ATP(NEN,Boston,MA),1μgミエリン塩基性タンパク、および0.1μg ROCK−1を含有する総体積100μLの96ウエルプレート中で実施される。テスト化合物は100%ジメチルスルホキシドに溶解され、適切な濃度に希釈され、そして反応へ添加される。ジメチルスルホキシドの最終濃度は0.5%をこえなかった。反応は室温で1時間行われる。反応はHClの7mLの添加によって停止され、P30メンブレン中へ移され、基質であるミエリン塩基性タンパクへ添加された〔33P〕ATPの量をカウント/分(c.p.m.)としてBetuPlateリーダー(Packard Instrument Co.,Mericlen,CT)中で読取られる。(すべての試薬は特記しない限りSigma Chemical Co.,St.Louis,MOから購入した。)阻害%はテスト化合物の存在なしで添加された量と比較した時の、テスト化合物の存在下の放射活性の取込み量によって測定される。
【0042】
また阻害活性は、Ridley,A.J.and A.Hall,Cell 70:389−399(1992)に記載されたのと本質的に同じように実施されるストレス線維生成の測定によって評価することができる。ヒト線維肉腫HT1080(CCL−121,American Type Culture Collection,Manassa,VA)細胞が10%ウシ胎児血清を補強したDelbeco修正イーグル培地(DMEM,Gibco)中の2.5×104細胞/ウエルにおいて6ウエル組織培養プレート(Costar)中の22×22mm#ガラスカバースリップ上にプレートされる。細胞は加湿された5%CO2雰囲気中に維持される。24時間後培養倍地を除去し、10%ウシ胎児血清なしの培地で交換され、細胞は追加の48時間培養される。テスト化合物は100%ジメチルスルホキシドに溶解され、適当な濃度に希釈され、ストレス線維の生成誘発前60分に培地へ添加される。ジメチルスルホキシドの最終濃度は0.25%をこえなかった。ストレス線維生成はリソフォスファチジン酸(1−オレイル−2−ヒドロキシ−Sn−グリセロール−3−フォスフェート,Avanti Polar−Lipids,Albaster,AI)を37℃において15分間0.1%脂肪酸不含ウシ血清アルブミンを含んでいるDelbeco修正イーグル培地へ10μM最終濃度に添加することによって誘発される。細胞はリン酸塩緩衝食塩水(PBS)中4%パラホルムアルデヒド(Poly Scientific,Bay Shore,NJ)で15分間固定される。細胞は次にPBS中3回洗浄され、そして次に40mMピペラジン−N,N’−ビス(2−エタンスルホン酸)、50mM(2−ヒドロキシエチル)ピペラジン−N’−(2−エタンスルホン酸)、0.1%トリトンX−100、75mM NaCl,mM MgCl2, 0.5mM EDTA,pH7.2を含んでいる溶液を用いて室温で2分間浸透性化される。細胞は各回PBS中5分間3回洗浄され、そして次にアクチンストレス線維がPBS中10単位/mLのローダミンファロイジン(Molecular Probes,Eugeme,OR)を用いて室温で60分間染色される。細胞はPBSで3回洗浄され、カバースリップがガラス顕微鏡スライド上に載置される。各スライド上のストレス線維陽性細胞のパーセントがニコンLabphto−2顕微鏡を用いて肉眼で決定された。少なくともプレートあたり100細胞がカウントされ、そして実験は2回実施された。阻害パーセントは、テスト化合物が存在しない場合のストレス線維陽性細胞の数と比較した時、テスト化合物存在下におけるストレス線維陽性線維の数をカウントすることによって測定される。
【0043】
上のプロトコールを用いて、ここに開示した化合物のすべてはRhoキナーゼ阻害活性を有することが決定される。
【0044】
本発明の化合物は、ルーチンな慣用化学的方法に従って、および/または以下に記載するように、商業的に入手できるか、またはルーチンな慣用化学的方法に従って製造することができる出発物質から製造することができる。化合物の製造のための一般的方法が以下に与えられ、そして代表的化合物の製造が実施例に特定的に例証されている。
【0045】
略号
ここで以下の略号が使用される時、それらは以下の意味を持つ。
Figure 2004528314
Figure 2004528314
【0046】
一般的製造方法
以下の一般的方法を記載するために使用した式中で、R1およひR2は水素もしくはメトキシであり、R3はメトキシエチル、シクロプロピル、4−フロロフェニルもしくは4−ピリジルであり、本発明の化合物I−VIを製造するために適宜選択される。
【0047】
一般法A:
【0048】
【化21】
Figure 2004528314
【0049】
THF/水中の化合物1および、そして酢酸カリウムの混合物を室温で一夜攪拌する。混合物へ水を加え、沈澱を生成させる。この沈澱を水で洗い、濾過し、そして高真空下乾燥して化合物3を得る。
【0050】
一般法B:
【0051】
【化22】
Figure 2004528314
【0052】
化合物3と、置換アミンもしくはアニリンの混合物を140℃へ2時間加熱する。混合物を室温へ冷却し、そしてエーテルで処理して沈澱を生成させるか、またはシリカゲルクロマトグラフィーによって精製する。沈澱の精製:沈澱を濾過し、エーテルで数回洗い、そして高真空下乾燥して生成物を得る。
【0053】
さらに考究することなく、当業者は以上の説明を利用して本発明をその全範囲において利用することができると信じられる。それ故以下の好ましい特定の具体例は単に例証であり、如何なる態様においても開示の残部の限定ではないと考えるべきである。
【0054】
以上および以下の実施例において、すべての温度は未補正の摂氏で与えられ、そして特記しない限りすべての部およびパーセントは重量による。
【0055】
2001年3月23日に出願された米国仮特許出願No.60/277,974および2001年8月29日に出願された米国仮特許出願No.60/315,338を含む、以前および以下に引用したすべての出願、特許および発表の全体の開示が参照としてここに取入れられる。
【0056】
〔実施例1〕
2−N−5’−アミノインダゾール−4−クロロ−6,7−ジメトキシキナゾリン
【0057】
【化23】
Figure 2004528314
【0058】
THF/水(2L/0.9L)中の2,4−ジクロロ−6,7−ジメトキシキナゾリン(Aldrich Chemical Co.,226g,0.874mol)、5−アミノインダゾール(130g,0.98mol)および酢酸カリウム(111.5g,1.14mo.)の混合物を室温で一夜攪拌する。水(2L)を混合物へ加え、沈澱を生成させる。この沈澱を水で洗い、濾過し、高真空下で乾燥し、生成物を灰色粉末として得る。
【0059】
〔実施例2〕
N−〔2−(2,4−ジクロロフェニル)−4−キナゾリニル〕−N−(1H−インダゾール−5−イル)アミンの製造
【0060】
【化24】
Figure 2004528314
【0061】
ブタノール(50mL)中の4−クロロ−2−フェニルキナゾリン(Aldrich Chemical Co.,7.2g,30mol)と、5−アミノインダゾール(3.99g,30mmol)の混合物を100℃へ一夜加熱する。溶媒を減圧除去した後、粗生成物をシリカゲルクロマトグラフィー(酢酸エチル/ヘキサン20%ないし80%勾配)よって精製し、実施例2の化合物(3.6g)を得る。HPLC/MS:(M+H)+338m/z,保持時間(HPLC/MS)=3.65分
【0062】
実施例3−6への一般的合成ルート
【0063】
【化25】
Figure 2004528314
【0064】
〔実施例3〕
N2−(4−フロロフェニル)−N4−(1H−インダゾール−5−イル)−6,7−ジメトキシ−2,4−キナゾリンジアミン
【0065】
【化26】
Figure 2004528314
【0066】
n−ブタノール(1mL)中の2−フロロ−N−(1H−インダゾール−5−イル)−6,7−ジメトキシ−4−キナゾリンアミン(0.1mmol)と、4−フロロアニリン(0.3mmol)の懸濁液を90℃で72時間振とうする。溶媒を蒸発し、残渣をHPLCによって精製し、純粋な生成物を得る。(M+H)+=431,RT(LC−MS)=2.92
【0067】
実施例3のための上に記載した方法を使用し、そして適切な出発物質に置換し、実施例4−6の化合物を同様に製造し、下の表1に要約する。
【0068】
【化27】
Figure 2004528314
【0069】
【表1】
Figure 2004528314
【0070】
上の実施例は本発明の一般的にまたは特定的に記載した反応剤および/または作業条件を以上の実施例に使用したそれらに置換することにより、類似の成功度をもってくり返すことができる。【Technical field】
[0001]
This application is a provisional application filed on March 23, 2001. No. 60 / 277,974 and U.S. patent application no. Claim the benefit of the filing date of 60 / 315,338.
[0002]
The present invention relates to compounds, derivatives thereof, their synthesis, and their use as Rho kinase inhibitors. These compounds of the invention are useful for tumor growth arrest, treatment of erectile dysfunction, and other indications mediated by Rho kinase, such as coronary heart disease.
[Background]
[0003]
A number of human and animal disease pathologies, including hypertension, erectile dysfunction, coronary circulatory disturbances, neurodegeneration disorders and cancer, can be directly linked to changes in the actin cytoskeleton. These diseases provide a serious unmet medical need. The actin cytoskeleton is composed of a network of actin filaments and actin-binding proteins found in all eukaryotic cells. In smooth muscle cells, actin cytoskeletal assembly and dissemination is the primary driving force responsible for smooth muscle contraction and relaxation. In non-smooth muscle cells, dynamic relocation of the actin cytoskeleton is specialized such as cell morphology, cell mobility, actin stress fiber formation, cell adhesion, and axonal contraction, phagocytosis or cell division Responsible for the regulation of cell function (Van Aelst, et al. Genes Dev 1997, 11, 295).
[0004]
The actin cytoskeleton is controlled by a family of proteins that are a subset of the Ras superfamily. This subset currently consists of RhoA to G and RhoG (collectively referred to as Rho), Rac1 and 2, Cdc42Hs and G25K and TC10 isoforms (Mackay, et al. J Bio Chem 1998, 273, 20685). These three proteins are GTP (guanine nucleotide triphosphate) binding proteins with intrinsic GTPase activity. They act as molecular switches and cycle between bound GDP (guanine nucleotide diphosphate) and the active GTP binding state. Using biochemical and genetic engineering, it became possible to assign functions to each family member. When activated, the Rho protein controls the formation of thick bundles of actin stress fibers and actin filaments and integrin clustering in the focal adhesion complex. When activated, the Rac protein controls the production of membrane undulations on the cell surface, and Cdc42 controls the production of filamentous feet. Together, these protein families play an important role in the control of key cell functions, including cell motility, axon guidance, cell division, and changes in cell morphology, shape and polarity.
[0005]
Depending on the type of cell and the activated receptor, Rho proteins can regulate various biological responses. In smooth muscle cells, Rho protein is responsible for calcium sensitization during smooth muscle contraction. In non-smooth muscle cells, Rho GTPase is lysophosphatidic acid (LPA), thrombin and thromboxane A.2Responsible for cellular responses to agonists such as (Fukuda, et al. Trends Polar Sci 2001, 22, 32). Agonist responses are linked through heterotrimeric G protein, Galpha12 or Galpha13, although other receptors may also be involved (Goetzl, et al. Cancer Res 1999, 59, 4732; Buhl, et al. J Biol Chem 1995, 270, 24681). When activated, Rho GTPase activates a number of downstream factors including PIP5-kinase, lowsekin, lophyrin, PKN, and Rho kinase isoforms ROCK-1 / ROK beta and ROCK-1 / ROK alpha (Mackay et al. J Biol Chem 1998, 273, 20687; Aspenstrom, Curr Opin Cell Biol 1999, 11, 95; Amano, et al. Exp Cell Res 2000, 261, 44).
[0006]
Rho kinase has been identified as a RhoA interacting protein isolated from bovine brain (Matsui, et al. Embo J 1996, 15, 2208). It is a member of the muscular dystrophy family of protein kinases and contains a serine / threonine domain at the amino terminus, a coiled domain at the central region, and a Rho interaction domain at the carboxy terminus (Amano, et al. Exp Cell Res). 2000, 261, 44). Its kinase activity is enhanced when bound to GTP-bound RhoA, and it reproduces the multiple activities of activated RhoA when introduced into cells. In smooth muscle cells, Rho kinase mediates calcium sensitization and smooth muscle contraction, and inhibition of Rho kinase blocks 5-HT and phenylephrine agonists induced by muscle contraction. When introduced into non-smooth muscle cells, Rho kinase induces stress fibrogenesis and is required for RhoA-mediated cell transformation (Sahai, et al. Curr Biol 1999, 9, 136). Rho kinase can be obtained by phosphorylation through myosin light chain (Somlyo, et al. J Physio (Lond) 2000, 522 pt2, 177), myosin light chain phosphatase binding subunit (Fukata, et al. J Cell Biol 1998, 141, 409) and a number of downstream proteins including LIM-kinase 2 (Sumi, et al. J BioChem 2001, 276, 670).
[0007]
Inhibition of Rho kinase in animal models has demonstrated numerous benefits of Rho kinase inhibitors for human disease. (+)-Trans-4- (1-aminoethyl)-(pyridin-4-ylaminocarbonyl) cyclohexanedihydrochloride-hydrate (WO00 / 078351, WO00 / 057913) and substituted isoquinolinesulfonyl compounds (EP00187371) There are several patents claiming as Rho kinase inhibitors having activity in animal models. These include hypertension (Vehata, et al. Nature 1997, 389, 990), arteriosclerosis (Retzer, et al. FEBS Lett 2000, 466, 70), restenosis (Eto, et al. Am J Physiol Heart Circ Physiol 2000, Negoro, et al., Biochem Biophys Res Commun 1999, 261, 211), cerebral ischemia (Uehata et al. Nature 1997, 389, 990); Seaholtz, et al. Circ Res 1999, 84, 1186; Hitomi, et al. Life Sci 2000, 67, 1929; Yamamoto, et al. J Cardiovas Pharmacol 2000, 35, 203), cerebral vasospasm (Sato, et al. Circ Res 2000, 87, 195; Kim, et al. Neurosurgery 2000, 46, 440), erectile dysfunction (Chitaley, et al. Nat. Med 2001, 7, 119), central nervous system disorders such as neurodegeneration and spinal cord disorders (Hara, et al. J Neurosurg 2000, 93, 94; Toshima, et al. Stroke 2000, 31, 245), Rho kinase. Neoplasia has been shown to inhibit tumor cell growth and metastasis (Itoh, et al. Nat Med 1999, 5, 221; Somlye, et al. Biochem Biophys Re Commun 2000, 269, 652), angiogenesis (Uchida, et al. Biochem Biophys Res Commun 2000, 269, 633; Gingras, et al. Biochem J 2000, 348 Pt 2,273), platelet aggregation (Klages, et al. J Cell Biol 1999, 144, 745; Retzer, et al. Cell Signal 2000, 12, 645) and leukocyte aggregation (Kawaguchi, et al. Eur J Pharmacol 2000, 403, 203; Sanchez-Madrid, et al. Embo J. 1999, 18, 501), arterial hemostasis disorder, asthma (Setoguchi, et al. Br J Pharmacol 20 Nakahara, et al. Eur J Pharmacol 2000, 389, 103), intraocular pressure regulation (Honjo, et al. Invest Ophthalmol Vis Sci 2001, 42, 137), and bone resorption (Chellaiah, et al. J Biol Chem 2000, 275, 11993; Zhang, et al. J Cell Sci 1995, 108, 2285).
[0008]
In patients, inhibition of Rho kinase has the benefit of controlling cerebral vasospasm and ischemia after subarachnoid hemorrhage (Pharmacia Japan 1995, 1470, 16).
DISCLOSURE OF THE INVENTION
[0009]
The compounds provided in the present invention and their derivatives are useful as Rho kinase inhibitors, so that hypertension, arteriosclerosis, restenosis, cerebral ischemia, cerebral vasospasm, nerve degeneration, bone marrow injury, breast, prostate, It has utility in the treatment of colorectal, ovarian, brain and lung cancers and their metastases, hemostatic disorders, asthma, glaucoma and osteoporosis.
[0010]
In addition, the compounds of the present invention are useful for treating erectile dysfunction, ie erectile dysfunction mediated by Rho kinase. Erectile dysfunction can be defined as the inability to obtain or maintain an erection appropriate for intercourse. WO 94/28902, U.S. Pat. S. P. 6,103,765 and U.S. Pat. S. P. See 6,124,461.
[0011]
The present invention provides compounds of the following formula:
[0012]
Embedded image
Figure 2004528314
[0013]
Embedded image
Figure 2004528314
[0014]
Embedded image
Figure 2004528314
[0015]
Embedded image
Figure 2004528314
[0016]
Embedded image
Figure 2004528314
[0017]
Embedded image
Figure 2004528314
[0018]
The present invention is also directed to pharmaceutically acceptable salts of formula I-VI. Suitable pharmaceutically acceptable salts are well known to those skilled in the art and are hydrochloric, hydrobromic, sulfuric, phosphoric, methanesulfonic, sulfonic, acetic, trifluoroacetic, malic, tartaric acid Salts of inorganic and organic acids such as citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, and mandelic acid. In addition, pharmaceutically acceptable salts include alkali cations (eg Li+, Na+Or K+), Alkaline earth cations (eg Mg++, Ca++, Or Ba++), Salts of inorganic bases such as salts containing ammonium cations, and triethylamine, N, N-diethylamine, N, N-dicyclohexylamine, pyridine, N, N-dimethylaminopyridine (DMAP), 1,4 -Diazabicyclo [2.2.2] octane (DABCO), 1,5-diazabicyclo [4.3.0] nonene-5 (DBN) and 1,8-diazabicyclo [5.4.0] undecene-7 (BBU) Organic base acid salts containing aliphatic and aromatic substituted ammonium and quaternary ammonium cations such as those generated from protonation or peralkylation of
[0019]
Many of the compounds of formula I-VI have asymmetric carbon atoms and therefore exist in racemic and optically active forms. Methods for separating enantiomeric and diastereomeric mixtures are well known to those skilled in the art. The invention includes any isolated racemic or optically active form of a compound of formula I-VI having Rho kinase inhibitory activity.
[0020]
The present invention also includes a pharmaceutical composition comprising a compound of formula I-VI and a physiologically acceptable carrier.
[0021]
The invention further comprises treating a Rho-kinase mediated indication by administering a compound of formula I-VI, or a pharmaceutical composition comprising a compound of formula I-VI. Thus, the present invention is directed against hypertension, arteriosclerosis, restenosis, cardiovascular diseases such as cerebral ischemia or spasticity, central nervous system disorders such as nerve degeneration and spinal cord injury, erectile dysfunction such as PDE-5. Treating patients who do not have a satisfactory response and cancers mediated by Rho kinase (eg tumor growth) by administering an effective amount of a compound of formula I, for example, to a host in need thereof. Including. Cancers and tumors mediated by Rho kinase include breast, large intestine, prostate, ovary, brain and lung cancers and their metastases.
[0022]
The compounds can be administered orally, topically, parenterally, by inhalation or spray, vaginally, enterally or sublingually in dosage unit formulations. Administration by injection includes intravenous, intraarterial, intramuscular, subcutaneous and parenteral injection and the use of infusion techniques. Dermal administration can include topical or transdermal administration. One or more compounds may be present with one or more non-toxic pharmaceutically acceptable carriers and, if desired, other active ingredients.
[0023]
Compositions intended for oral use can be prepared according to any suitable known method for the manufacture of pharmaceutical compositions. Such compositions can contain one or more agents selected from the group consisting of diluents, sweeteners, flavoring agents, colorants and preservatives to provide a dosage form that can be taken. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable adjuvants which are suitable for the manufacture of tablets. These adjuvants include, for example, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate, granulating and disintegrating agents such as corn starch or alginic acid, and binders such as magnesium stearate, stearin Acid or talc. Tablets can be uncoated or delayed disintegration and absorption in the gastrointestinal tract, thereby allowing them to be coated for extended periods of time. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be used. These compounds can also be prepared in a solid immediate release form.
[0024]
Formulations for oral use can also be provided as hard gelatin capsules, in which case the active ingredient is mixed with an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin, or the active ingredient is water or an oil medium For example, soft gelatin capsules mixed with peanut oil, liquid paraffin or olive oil.
[0025]
Aqueous suspensions containing the active ingredients in admixture with adjuvants suitable for the manufacture of aqueous suspensions can also be used. Such adjuvants are suspending agents such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum arabic, and dispersing or wetting agents are natural phosphatides such as lecithin or alkylene oxides of fatty acids Condensates such as polyoxyethylene stearate, or alkylene oxide condensates with long chain fatty alcohols such as heptadecaethyleneoxycetanol, or alkylenes of partial esters derived from fatty acids and hexitol such as polyoxyethylene sorbitol monooleate Oxide condensates or alkylene oxide condensates of partial esters derived from fatty acids and hexitol anhydrides such as poly It can be a carboxymethyl sorbitan monooleate. Aqueous suspensions or one or more preservatives such as ethyl p-hydroxybenzoate or n-propyl ester, one or more colorants, one or more flavoring agents, and one such as sucrose or saccharin The above sweeteners can be included.
[0026]
Dispersible powders or granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional adjuvants such as sweetening, flavoring and coloring agents may also be present.
[0027]
The compounds can be formulated in a non-aqueous liquid formulation, for example by suspending the active ingredient in a vegetable oil such as arachis oil, olive oil, sesame oil or peanut oil, or in a mineral oil such as liquid paraffin. It may be a turbid liquid. Oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening and flavoring agents such as those described above can be added to provide an easy to take oral formulation. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
[0028]
The compounds of the present invention may be prepared by methods known to those skilled in the art (see, for example, Chien; “Transdermal Controlled Systems Medicines”; Marcel Dekker, Inc .; 1987. Lipp et al. WO 94/04157, March 3, 1994). It can also be administered transdermally. For example, a solution or suspension of a compound of formula I-VI in a suitable volatile solvent optionally containing a penetration enhancer is combined with additional additives known to those skilled in the art such as matrix materials and bactericides. Can be matched. After sterilization, the resulting mixture can be formulated into dosage forms according to known procedures. In addition, in treatment with emulsifiers and water, solutions or suspensions of compounds of formula I-VI can be formulated into lotions or salves.
[0029]
Suitable solvents for processing into transdermal delivery systems are known to those skilled in the art and include lower alcohols such as ethanol or propyl alcohol, lower ketones such as acetone, lower carboxylic acid esters such as ethyl acetate, tetrahydrofuran. Polar ethers such as hexane, cyclohexane or benzene, or halogenated hydrocarbons such as dichloromethane, chloroform, trichlorotrifluoroethane or trichlorofluoroethane. Suitable solvents can include a mixture of one or more materials selected from lower alcohols, lower ketones, lower carboxylic esters, polar ethers, lower hydrocarbons and halogenated hydrocarbons.
[0030]
Penetration enhancers suitable for transdermal delivery systems are known to those skilled in the art and are saturated or unsaturated such as monohydroxy or polyhydroxy alcohols such as ethanol, propylene glycol or benzyl alcohol, lauryl alcohol or cetyl alcohol. Saturation C8-C18Saturated or unsaturated C such as fatty alcohol, stearic acid8-C18Up to 24 such as fatty acid, acetic acid, caproic acid, lauric acid, myristic acid, stearic acid or palmitic acid, methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t-butyl or monoglycerin ester Saturated or unsaturated fatty acid esters having carbon atoms, or diesters of saturated or unsaturated fatty acids having up to 24 carbon atoms such as diisopropyl adipate, diisobutyl adipate, diisopropyl sebacate, diisopropyl maleate or diisopropyl fumarate. Additional penetration enhancers include phosphatidyl derivatives such as lecithin or cephalin, terpenes, amides, ketones, urea and derivatives thereof, and ethers such as dimethyl isosorbide and diethylene glycol monoethyl ether. Suitable penetration enhancers are monohydroxy or polyhydroxy alcohols, saturated or unsaturated C8-C18Fatty alcohol, saturated or unsaturated C8-C18From fatty acids, saturated or unsaturated fatty acid esters having up to 24 carbon atoms, diesters of saturated or unsaturated dicarboxylic acids having up to 24 carbons in total, phosphatidyl derivatives, terpenes, amides, ketones, ureas and their derivatives and ethers It may contain a mixture of one or more selected materials.
[0031]
Suitable bonding materials for transdermal delivery systems are known to those skilled in the art and include polyacrylates, silicones, polyurethanes, block copolymers, styrene butadiene copolymers, natural and synthetic rubbers. Cellulose ethers, derivatized polyethylene and silicates can also be used as matrix components. Additional additives such as viscous resins or oils can be added to increase the viscosity of the matrix.
[0032]
The pharmaceutical composition of the present invention may be in the form of an O / W emulsion. The oily phase may be a vegetable oil, for example arachis oil or olive oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifiers are natural gums such as gum arabic or tragacanth, natural phosphatides such as soy lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides such as sorbitan monooleate, and condensates of said partial esters with ethylene oxide such as polyoxy It can be ethylene sorbitan monooleate. The emulsion can contain sweetening and flavoring agents.
[0033]
Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations can contain irritating agents, preservatives, flavoring and coloring agents.
[0034]
The compounds can also be administered in the form of rectal or vaginal suppositories for drug administration. These compositions are solid at room temperature, but are liquid at rectal or vaginal temperatures and are mixed with a suitable nonirritating adjuvant that melts in the rectum or vagina to release the drug. Can be prepared. Such materials include cocoa butter and polyethylene glycol.
[0035]
Furthermore, for the treatment of erectile dysfunction, the pharmaceutical composition may be in any form suitable for administration to the penis by injection into the clitoral corpus cavernosum or transurethral administration or by topical application to the urethra. Can take. For injections into the clitoral corpus cavernosum, the pharmaceutical composition is preferably a saline solution. Preferably the pharmaceutical composition is in a form suitable for transurethral administration, and in this case the composition is typically in the form of a solution, ointment or suppository. Typically, the pharmaceutical composition is administered 1 to 50 minutes, preferably 10 to 20 minutes before the start of sexual intercourse.
[0036]
In all therapies of use disclosed herein for compounds of formula I, the daily oral dosage regimen will preferably be from 0.01 to 200 mg / kg of total body weight.
[0037]
The daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injection, and by use of infusion techniques will preferably be from 0.01 to 200 mg per kg body weight. Daily vaginal therapy will preferably be from 0.01 to 200 mg / kg of total body weight. Daily topical therapy will preferably be from 0.01 to 200 mg / kg of total body weight administered 1 to 4 times per day. The transdermal concentration will preferably be that required to maintain a daily dosage of 0.1 to 200 mg / kg. Daily inhalation therapy will preferably be from 0.01 to 10 mg / kg of total body weight.
[0038]
One skilled in the art will recognize that the particular mode of administration will depend on various factors that are routinely considered when administering a therapeutic agent. However, the specific dose level for a given patient depends on the activity of the specific compound used, patient age, patient weight, patient general health, patient sex, patient diet, administration time, administration It will also be appreciated that it will depend on a variety of factors including route, excretion rate, drug combination, and the severity of the condition being treated. In addition, those skilled in the art will use conventional treatment tests to determine the optimal course of treatment, ie the mode of treatment and the number of daily doses of the compound of formula I or pharmaceutically acceptable salt administered over a limited number of days. It will be appreciated that this can be verified.
[0039]
The compounds and compositions exhibit Rho kinase inhibitory activity and are therefore useful in the treatment of the indications listed above, eg, indications mediated by Rho kinase. An indication mediated by Rho kinase means a disease or condition whose progression proceeds at least in part via the Rho pathway.
[0040]
Rho kinase inhibitory activity, such as ROCK-1 inhibition, can be evaluated as follows.
[0041]
Amino acids 27-530, the kinase domain of human ROCK, are isolated from Sf9 insect cells as a glutathione S-transferase enzyme fusion protein. This protein is partially purified by glutathione sepharose 4B (Pharmacia Biotech, NJ) affinity purification. The reaction was 50 mM N- (2-hydroxyethyl) piperazine-N ′-(2-ethanesulfonic acid) pH 7.5, 5 μM MgCl2, 1 mM dithiothreitol, 6 μM ATP, 0.2 μCi [33Performed in 96 well plates with a total volume of 100 μL containing P] ATP (NEN, Boston, Mass.), 1 μg myelin basic protein, and 0.1 μg ROCK-1. The test compound is dissolved in 100% dimethyl sulfoxide, diluted to the appropriate concentration and added to the reaction. The final concentration of dimethyl sulfoxide did not exceed 0.5%. The reaction is carried out at room temperature for 1 hour. The reaction was stopped by the addition of 7 mL of HCl, transferred into a P30 membrane and added to the substrate myelin basic protein [33The amount of P] ATP is read in a BetaPlate reader (Packard Instrument Co., Mericlen, CT) as counts per minute (cpm). (All reagents were purchased from Sigma Chemical Co., St. Louis, MO unless otherwise specified.) Percent inhibition is the radioactivity in the presence of test compound as compared to the amount added without the presence of test compound. Measured by the amount of uptake.
[0042]
Inhibitory activity is also shown in Ridley, A. et al. J. et al. and A. Hall, Cell 70: 389-399 (1992) and can be assessed by measurement of stress fiber production performed essentially as described. Human fibrosarcoma HT1080 (CCL-121, American Type Culture Collection, Manassa, Va.) 2.5 × 10 × in Delbeco's modified Eagle medium (DMEM, Gibco) supplemented with 10% fetal bovine serum.FourCells are plated onto 22 × 22 mm # glass coverslips in 6 well tissue culture plates (Costar) in cells / well. Cells are humidified 5% CO2Maintained in atmosphere. After 24 hours, the culture medium is removed and replaced with medium without 10% fetal calf serum and the cells are cultured for an additional 48 hours. The test compound is dissolved in 100% dimethyl sulfoxide, diluted to an appropriate concentration, and added to the medium 60 minutes before induction of stress fiber formation. The final concentration of dimethyl sulfoxide did not exceed 0.25%. Stress fiber formation was achieved by lysophosphatidic acid (1-oleyl-2-hydroxy-Sn-glycerol-3-phosphate, Avanti Polar-Lipids, Albaster, AI) at 37 ° C. for 15 minutes with 0.1% fatty acid-free cattle. Induced by adding 10 μM final concentration to Delbeco modified Eagle medium containing serum albumin. Cells are fixed with 4% paraformaldehyde (Poly Scientific, Bay Shore, NJ) in phosphate buffered saline (PBS) for 15 minutes. The cells are then washed 3 times in PBS and then 40 mM piperazine-N, N′-bis (2-ethanesulfonic acid), 50 mM (2-hydroxyethyl) piperazine-N ′-(2-ethanesulfonic acid). 0.1% Triton X-100, 75 mM NaCl, mM MgCl2, And is permeabilized for 2 minutes at room temperature using a solution containing 0.5 mM EDTA, pH 7.2. Cells are washed 3 times for 5 minutes each in PBS and then actin stress fibers are stained with 10 units / mL rhodamine phalloidin (Molecular Probes, Eugene, OR) in PBS for 60 minutes at room temperature. Cells are washed 3 times with PBS and a coverslip is placed on a glass microscope slide. The percentage of stress fiber positive cells on each slide was determined visually using a Nikon Labphto-2 microscope. At least 100 cells per plate were counted and the experiment was performed twice. The percent inhibition is measured by counting the number of stress fiber positive fibers in the presence of the test compound when compared to the number of stress fiber positive cells in the absence of the test compound.
[0043]
Using the above protocol, all of the compounds disclosed herein are determined to have Rho kinase inhibitory activity.
[0044]
The compounds of the present invention are prepared according to routine conventional chemical methods and / or from starting materials that are commercially available or can be prepared according to routine conventional chemical methods, as described below. Can do. General methods for the preparation of compounds are given below and the preparation of representative compounds is specifically illustrated in the examples.
[0045]
Abbreviation
Here, when the following abbreviations are used, they have the following meanings.
Figure 2004528314
Figure 2004528314
[0046]
General manufacturing method
In the formula used to describe the following general method:1R2Is hydrogen or methoxy and RThreeIs methoxyethyl, cyclopropyl, 4-fluorophenyl or 4-pyridyl, and is appropriately selected for producing the compounds I-VI of the present invention.
[0047]
General method A:
[0048]
Embedded image
Figure 2004528314
[0049]
A mixture of Compound 1 in THF / water and potassium acetate is stirred overnight at room temperature. Water is added to the mixture to form a precipitate. The precipitate is washed with water, filtered and dried under high vacuum to give compound 3.
[0050]
General method B:
[0051]
Embedded image
Figure 2004528314
[0052]
A mixture of compound 3 and a substituted amine or aniline is heated to 140 ° C. for 2 hours. The mixture is cooled to room temperature and treated with ether to form a precipitate or purified by silica gel chromatography. Purification of the precipitate: The precipitate is filtered, washed several times with ether and dried under high vacuum to give the product.
[0053]
Without further consideration, it is believed that one skilled in the art, using the above description, can utilize the present invention in its full scope. The following preferred specific embodiments are therefore to be considered merely illustrative and not limiting of the remainder of the disclosure in any way.
[0054]
In the examples above and below, all temperatures are given in uncorrected degrees Celsius, and all parts and percentages are by weight unless otherwise specified.
[0055]
US provisional patent application no. 60 / 277,974 and US provisional patent application no. The entire disclosures of all applications, patents and publications cited above and below, including 60 / 315,338, are hereby incorporated by reference.
[0056]
[Example 1]
2-N-5'-aminoindazole-4-chloro-6,7-dimethoxyquinazoline
[0057]
Embedded image
Figure 2004528314
[0058]
2,4-Dichloro-6,7-dimethoxyquinazoline (Aldrich Chemical Co., 226 g, 0.874 mol), 5-aminoindazole (130 g, 0.98 mol) and acetic acid in THF / water (2 L / 0.9 L) A mixture of potassium (111.5 g, 1.14 mo.) Is stirred overnight at room temperature. Water (2 L) is added to the mixture and a precipitate is formed. The precipitate is washed with water, filtered and dried under high vacuum to give the product as a gray powder.
[0059]
[Example 2]
Preparation of N- [2- (2,4-dichlorophenyl) -4-quinazolinyl] -N- (1H-indazol-5-yl) amine
[0060]
Embedded image
Figure 2004528314
[0061]
A mixture of 4-chloro-2-phenylquinazoline (Aldrich Chemical Co., 7.2 g, 30 mol) and 5-aminoindazole (3.99 g, 30 mmol) in butanol (50 mL) is heated to 100 ° C. overnight. After removing the solvent under reduced pressure, the crude product is purified by silica gel chromatography (ethyl acetate / hexane 20% to 80% gradient) to give the compound of Example 2 (3.6 g). HPLC / MS: (M + H)+338 m / z, retention time (HPLC / MS) = 3.65 minutes
[0062]
General synthetic route to Examples 3-6
[0063]
Embedded image
Figure 2004528314
[0064]
Example 3
N2- (4-Fluorophenyl) -N4- (1H-indazol-5-yl) -6,7-dimethoxy-2,4-quinazolinediamine
[0065]
Embedded image
Figure 2004528314
[0066]
2-Fluoro-N- (1H-indazol-5-yl) -6,7-dimethoxy-4-quinazolinamine (0.1 mmol) and 4-fluoroaniline (0.3 mmol) in n-butanol (1 mL) The suspension is shaken at 90 ° C. for 72 hours. The solvent is evaporated and the residue is purified by HPLC to give the pure product. (M + H)+= 431, RT (LC-MS) = 2.92
[0067]
Using the method described above for Example 3 and substituting the appropriate starting materials, the compounds of Examples 4-6 are prepared analogously and summarized in Table 1 below.
[0068]
Embedded image
Figure 2004528314
[0069]
[Table 1]
Figure 2004528314
[0070]
The above examples can be repeated with similar success by substituting the generally and specifically described reactants and / or operating conditions of the present invention for those used in the previous examples.

Claims (12)

下記式IないしVIのいずれかの化合物または薬剤的に許容し得るそれらの塩:
Figure 2004528314
Figure 2004528314
Figure 2004528314
Figure 2004528314
Figure 2004528314
Figure 2004528314
A compound of any of the following formulas I to VI or a pharmaceutically acceptable salt thereof:
Figure 2004528314
Figure 2004528314
Figure 2004528314
Figure 2004528314
Figure 2004528314
Figure 2004528314
式Iの請求項1の化合物:
Figure 2004528314
The compound of claim 1 of formula I:
Figure 2004528314
式IIの請求項1の化合物:
Figure 2004528314
The compound of claim 1 of formula II:
Figure 2004528314
式III の請求項1の化合物:
Figure 2004528314
The compound of claim 1 of formula III:
Figure 2004528314
式IVの請求項1の化合物:
Figure 2004528314
The compound of claim 1 of formula IV:
Figure 2004528314
式Vの請求項1の化合物:
Figure 2004528314
The compound of claim 1 of formula V:
Figure 2004528314
式VIの請求項1の化合物:
Figure 2004528314
The compound of claim 1 of formula VI:
Figure 2004528314
請求項1の化合物をそれを必要とする宿主へ投与することよりなる、Rhoキナーゼによって仲介される適応症を処置する方法。A method of treating an indication mediated by Rho kinase, comprising administering the compound of claim 1 to a host in need thereof. 請求項1の化合物をそれを必要とする宿主へ投与することよりなる、高血圧、動脈硬化、再狭窄、脳虚血症、脳血管痙れん、神経退化、骨髄傷害、乳房、大腸、前立腺、卵巣、脳もしくは肺のがん、止血障害、喘息、緑内障、骨粗しょう症、または勃起不全を処置する方法。2. Hypertension, arteriosclerosis, restenosis, cerebral ischemia, cerebrovascular spasm, nerve degeneration, bone marrow injury, breast, large intestine, prostate, ovary comprising administering the compound of claim 1 to a host in need thereof To treat cancer of the brain, lung, hemostasis, asthma, glaucoma, osteoporosis, or erectile dysfunction. 宿主はヒトである請求項8の方法。The method of claim 8, wherein the host is a human. 宿主はヒトである請求項9の方法。The method of claim 9, wherein the host is a human. (a)塩基の存在下式1の化合物を式2の化合物を反応させて式3の化合物を製造し、
Figure 2004528314
(b)場合により式3の化合物をR3NH2またはAr2NH2と反応させて式4の化合物を製造する
Figure 2004528314
(式中R3はCH3O−CH2CH2−またはシクロプロピル、Ar2は4−フロロフェニルまたはピリジルである。)ことを特徴とする請求項1の化合物を製造する方法。
(A) reacting a compound of formula 1 with a compound of formula 2 in the presence of a base to produce a compound of formula 3;
Figure 2004528314
(B) optionally reacting a compound of formula 3 with R 3 NH 2 or Ar 2 NH 2 to produce a compound of formula 4
Figure 2004528314
(Wherein R 3 is CH 3 O—CH 2 CH 2 — or cyclopropyl, and Ar 2 is 4-fluorophenyl or pyridyl).
JP2002576235A 2001-03-23 2002-03-22 Rho kinase inhibitor Expired - Fee Related JP4320705B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US27797401P 2001-03-23 2001-03-23
US31533801P 2001-08-29 2001-08-29
PCT/US2002/008660 WO2002076977A2 (en) 2001-03-23 2002-03-22 Rho-kinase inhibitors

Publications (2)

Publication Number Publication Date
JP2004528314A true JP2004528314A (en) 2004-09-16
JP4320705B2 JP4320705B2 (en) 2009-08-26

Family

ID=26958818

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2002576235A Expired - Fee Related JP4320705B2 (en) 2001-03-23 2002-03-22 Rho kinase inhibitor

Country Status (19)

Country Link
US (3) US20030087919A1 (en)
EP (1) EP1370552B1 (en)
JP (1) JP4320705B2 (en)
AR (1) AR035792A1 (en)
AT (1) ATE353889T1 (en)
AU (1) AU2002245709A1 (en)
CA (1) CA2441501C (en)
CY (1) CY1108041T1 (en)
DE (1) DE60218138T2 (en)
DK (1) DK1370552T3 (en)
ES (1) ES2280517T3 (en)
HK (1) HK1061029A1 (en)
MX (1) MXPA03008659A (en)
MY (1) MY142915A (en)
PE (1) PE20020958A1 (en)
PT (1) PT1370552E (en)
TW (1) TWI336328B (en)
UY (1) UY27225A1 (en)
WO (1) WO2002076977A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007142323A1 (en) * 2006-06-08 2007-12-13 Ube Industries, Ltd. Novel indazole derivative having spiro ring structure in side chain
JP2008534518A (en) * 2005-03-25 2008-08-28 サーフェイス ロジックス,インコーポレイティド Improved pharmacokinetic compound
JP2012229240A (en) * 2006-12-08 2012-11-22 Irm Llc Compound and composition as protein kinase inhibitors
WO2017150174A1 (en) * 2016-03-04 2017-09-08 株式会社シンスター・ジャパン Pharmaceutical composition containing 2,4-diaminoquinazoline derivative or salt thereof as active ingredient, and 2,4-diaminoquinazoline derivative having specific structure

Families Citing this family (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
WO2000041698A1 (en) 1999-01-13 2000-07-20 Bayer Corporation φ-CARBOXY ARYL SUBSTITUTED DIPHENYL UREAS AS p38 KINASE INHIBITORS
PT1478358E (en) 2002-02-11 2013-09-11 Bayer Healthcare Llc Sorafenib tosylate for the treatment of diseases characterized by abnormal angiogenesis
US7645878B2 (en) * 2002-03-22 2010-01-12 Bayer Healthcare Llc Process for preparing quinazoline Rho-kinase inhibitors and intermediates thereof
KR101092048B1 (en) 2002-08-29 2011-12-12 산텐 세이야꾸 가부시키가이샤 REMEDY FOR GLAUCOMA COMPRISING Rho KINASE INHIBITOR AND PROSTAGLANDINS
GB0307333D0 (en) * 2003-03-29 2003-05-07 Astrazeneca Ab Therapeutic agent
EP1616577B1 (en) * 2003-04-18 2011-09-21 Senju Pharmaceutical Co., Ltd. 2-chloro-6,7-dimethoxy-N-5 [5-(1)H- indazolyl]quinazoline-4-amine, N-(1-benzyl-4-piperidinyl)-1H-indazole-5-amine dihydrochloride, 4-[2-(2,3,4,5,6-pentafluorophenyl)acryloyl]cinnamic acid and fasudil hydrochloride for use in repairing corneal perception.
PL1626714T3 (en) 2003-05-20 2007-12-31 Bayer Healthcare Llc Diaryl ureas for diseases mediated by pdgfr
CN1856469B (en) 2003-07-23 2013-03-06 拜耳医药保健有限责任公司 Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
WO2005035506A1 (en) 2003-10-15 2005-04-21 Ube Industries, Ltd. Novel indazole derivative
WO2006094626A1 (en) 2005-03-07 2006-09-14 Bayer Healthcare Ag Pharmaceutical composition comprising an omega- carboxyaryl substituted diphenyl urea for the treatment of cancer
CA2621181C (en) 2005-08-30 2011-04-19 Asahi Kasei Pharma Corporation 5-isoquinolinesulfonamide compounds
WO2008054599A2 (en) * 2006-09-27 2008-05-08 Surface Logix, Inc. Rho kinase inhibitors
RS53588B1 (en) 2006-12-08 2015-02-27 Irm Llc Compounds and compositions as protein kinase inhibitors
US8415372B2 (en) 2007-02-27 2013-04-09 Asahi Kasei Pharma Corporation Sulfonamide compound
CN101622243B (en) 2007-02-28 2013-12-04 旭化成制药株式会社 Sulfonamide derivative
CN101730690B (en) 2007-07-02 2013-01-16 旭化成制药株式会社 Sulfonamide compound, and crystal thereof
US9248125B2 (en) 2007-08-29 2016-02-02 Senju Pharmaceutical Co., Ltd. Agent for promoting corneal endothelial cell adhesion
EP3904505A1 (en) 2009-04-22 2021-11-03 Viacyte, Inc. Cell compositions derived from dedifferentiated reprogrammed cells
US9109245B2 (en) 2009-04-22 2015-08-18 Viacyte, Inc. Cell compositions derived from dedifferentiated reprogrammed cells
CN105861446B (en) 2009-10-16 2021-10-01 斯克里普斯研究所 Induction of pluripotent cells
EP3255142A1 (en) 2009-10-19 2017-12-13 Cellular Dynamics International, Inc. Cardiomyocyte production
ES2670842T3 (en) 2010-06-15 2018-06-01 Cellular Dynamics International, Inc. Generation of induced pluripotent stem cells from small volumes of peripheral blood
KR102160721B1 (en) 2010-12-22 2020-09-29 페이트 세러퓨틱스, 인코포레이티드 Cell culture platform for single cell sorting and enhanced reprogramming of iPSCs
AU2012225735B2 (en) 2011-03-04 2016-03-10 Glaxosmithkline Intellectual Property Development Limited Amino-quinolines as kinase inhibitors
EP2694644B1 (en) 2011-03-30 2018-01-24 Cellular Dynamics International, Inc. Priming of pluripotent stem cells for neural differentiation
EP2732029B1 (en) 2011-07-11 2019-01-16 FUJIFILM Cellular Dynamics, Inc. Methods for cell reprogramming and genome engineering
TWI547494B (en) 2011-08-18 2016-09-01 葛蘭素史克智慧財產發展有限公司 Amino quinazolines as kinase inhibitors
WO2013086236A2 (en) 2011-12-06 2013-06-13 Advanced Cell Technology, Inc. Method of directed differentiation producing corneal endothelial cells, compositions thereof, and uses thereof
EP2799537B1 (en) 2011-12-28 2021-09-22 Kyoto Prefectural Public University Corporation Normalization of culture of corneal endothelial cells
EP2628482A1 (en) 2012-02-17 2013-08-21 Academisch Medisch Centrum Rho kinase inhiitors for use in the treatment of neuroblastoma
EP2826855B1 (en) 2012-03-15 2018-08-29 iHeart Japan Corporation Myocardial sheet
EP2837681B1 (en) 2012-04-06 2018-02-21 Kyoto University Method for inducing erythropoietin-producing cell
TWI592417B (en) 2012-09-13 2017-07-21 葛蘭素史克智慧財產發展有限公司 Prodrugs of amino quinazoline kinase inhibitor
KR20150118152A (en) 2013-02-21 2015-10-21 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Quinazolines as kinase inhibitors
US8859286B2 (en) 2013-03-14 2014-10-14 Viacyte, Inc. In vitro differentiation of pluripotent stem cells to pancreatic endoderm cells (PEC) and endocrine cells
US10266807B2 (en) 2013-04-03 2019-04-23 FUJIFILM Cellular Dynamics, Inc. Methods and compositions for culturing endoderm progenitor cells in suspension
EP2985344B1 (en) 2013-04-12 2018-07-18 Kyoto University Method for inducing alveolar epithelium progenitor cells
AU2014279065B2 (en) 2013-06-11 2020-04-09 Astellas Pharma Inc. Method for producing renal precursor cells, and drug containing renal precursor cells
WO2015016371A1 (en) 2013-07-30 2015-02-05 京都府公立大学法人 Corneal endothelial cell marker
US9796962B2 (en) 2013-08-07 2017-10-24 Kyoto University Method for generating pancreatic hormone-producing cells
MY184219A (en) 2013-09-05 2021-03-26 Univ Kyoto New method for inducing dopamine-producing neural precursor cells
RU2712967C2 (en) 2013-10-31 2020-02-03 Киото Прифекчурал Паблик Юниверсити Корпорэйшн Therapeutic drug for diseases related to endoplasmic reticulum cell death in corneal endothelium
MX2016006915A (en) 2013-11-27 2017-01-23 Kyoto Prefectural Public Univ Corp Application of laminin to corneal endothelial cell culture.
US11268069B2 (en) 2014-03-04 2022-03-08 Fate Therapeutics, Inc. Reprogramming methods and cell culture platforms
JP6730608B2 (en) 2014-05-21 2020-07-29 国立大学法人京都大学 Method for producing pancreatic blast cells and therapeutic agent for pancreatic diseases containing pancreatic blast cells
US10711249B2 (en) 2014-12-26 2020-07-14 Kyoto University Method for inducing hepatocytes
US11441126B2 (en) 2015-10-16 2022-09-13 Fate Therapeutics, Inc. Platform for the induction and maintenance of ground state pluripotency
CN108463548B (en) 2015-10-30 2023-04-18 加利福尼亚大学董事会 Method for producing T cell from stem cell and immunotherapy method using the T cell
US20190083543A1 (en) 2016-02-15 2019-03-21 Kyoto Prefectural Public University Corporation Human functional corneal endothelial cell and application thereof
CA3021628A1 (en) 2016-04-22 2017-10-26 Kyoto University Method for producing dopamine-producing neural precursor cells
CN109844102B (en) 2016-08-16 2023-11-10 富士胶片细胞动力股份有限公司 Method for differentiating pluripotent cells
US11821007B2 (en) 2017-05-25 2023-11-21 Kyoto University Method for inducing differentiation of intermediate mesodermal cell to renal progenitor cell, and method for inducing differentiation of pluripotent stem cell to renal progenitor cell
EP3708662A4 (en) 2017-11-10 2021-08-25 Regenesis Science Co., Ltd. Method for producing cultured cell, and method for producing therapeutic agent for spinal cord injury disease
US11739326B2 (en) 2017-11-14 2023-08-29 Massachusetts Eye And Ear Infirmary RUNX1 inhibition for treatment of proliferative vitreoretinopathy and conditions associated with epithelial to mesenchymal transition
US11446273B2 (en) 2017-12-21 2022-09-20 Santen Pharmaceutical Co., Ltd. Medicament comprising combination of sepetaprost and Rho-associated coiled-coil containing protein kinase inhibitor
US20220267736A1 (en) 2017-12-28 2022-08-25 Kaneka Corporation Pluripotent stem cell aggregation suppressor
JP7349911B2 (en) 2017-12-28 2023-09-25 株式会社カネカ Cell aggregation promoter
JP7257333B2 (en) 2017-12-28 2023-04-13 株式会社カネカ cell aggregation inhibitor
WO2019160148A1 (en) 2018-02-19 2019-08-22 大日本住友製薬株式会社 Cell aggregate, mixture of cell aggregates, and method for preparing same
JP7357369B2 (en) 2018-07-23 2023-10-06 国立大学法人京都大学 Novel renal progenitor cell marker and method for enriching renal progenitor cells using it
US20210292766A1 (en) 2018-08-29 2021-09-23 University Of Massachusetts Inhibition of Protein Kinases to Treat Friedreich Ataxia
JPWO2020045642A1 (en) 2018-08-31 2021-08-12 学校法人同志社 Compositions and Methods for Preserving or Culturing Eye Cells
CN113015429A (en) 2018-10-02 2021-06-22 学校法人同志社 Method and container for preserving corneal endothelial cells
EP3900787A4 (en) 2018-12-21 2022-02-23 Kyoto University Lubricin-localized cartilage-like tissue, method for producing same and composition comprising same for treating articular cartilage damage
WO2020193802A1 (en) 2019-03-28 2020-10-01 Fundación De La Comunidad Valenciana Centro De Investigación Príncipe Felipe Polymeric conjugates and uses thereof
JPWO2020203538A1 (en) 2019-03-29 2020-10-08
JPWO2020230832A1 (en) 2019-05-15 2020-11-19
CN115427551A (en) 2020-02-27 2022-12-02 京都府公立大学法人 Human functional corneal endothelial cell and application thereof
WO2022149616A1 (en) 2021-01-08 2022-07-14 国立大学法人京都大学 Medium for culturing and expanding nephron progenitor cells, method for culturing and expanding nephron progenitor cells, and method for producing renal organoids
EP4274578A1 (en) 2021-01-11 2023-11-15 Incyte Corporation Combination therapy comprising jak pathway inhibitor and rock inhibitor
KR20230165846A (en) 2021-04-07 2023-12-05 후지필름 셀룰러 다이내믹스, 인코포레이티드 Dopaminergic progenitor cells and methods of use
EP4334437A1 (en) 2021-05-03 2024-03-13 Astellas Institute for Regenerative Medicine Methods of generating mature corneal endothelial cells
CA3222761A1 (en) 2021-06-10 2022-12-15 Ajinomoto Co., Inc. Method for producing mesenchymal stem cells
WO2023017848A1 (en) 2021-08-11 2023-02-16 国立大学法人京都大学 Method for producing renal interstitial progenitor cells, erythropoietin-producing cells, and method for producing renin-producing cells
AU2022343749A1 (en) 2021-09-13 2024-03-28 FUJIFILM Cellular Dynamics, Inc. Methods for the production of committed cardiac progenitor cells
WO2024073776A1 (en) 2022-09-30 2024-04-04 FUJIFILM Cellular Dynamics, Inc. Methods for the production of cardiac fibroblasts

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06336481A (en) * 1992-12-10 1994-12-06 Zeneca Ltd Quinazoline derivative, its preparation and medicinal composition having anticancerous action and containing same
WO1995015758A1 (en) * 1993-12-10 1995-06-15 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit csf-1r receptor tyrosine kinase
WO1996039145A1 (en) * 1995-06-06 1996-12-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Protein tyrosine kinase aryl and heteroaryl quinazoline compounds having selective inhibition of her-2 autophosphorylation properties
WO1999006396A1 (en) * 1997-07-29 1999-02-11 Warner-Lambert Company Irreversible bicyclic inhibitors of tyrosine kinases
WO1999035146A1 (en) * 1998-01-12 1999-07-15 Glaxo Group Limited Bicyclic heteroaromatic compounds as protein tyrosine kinase inhibitors
WO1999035132A1 (en) * 1998-01-12 1999-07-15 Glaxo Group Limited Heterocyclic compounds
JPH11508906A (en) * 1995-07-13 1999-08-03 グラクソ、グループ、リミテッド Heterocyclic compounds and pharmaceutical compositions containing them
GB2345486A (en) * 1999-01-11 2000-07-12 Glaxo Group Ltd Heteroaromatic protein tyrosine kinase inhibitors
WO2000044728A1 (en) * 1999-01-27 2000-08-03 Pfizer Products Inc. Substituted bicyclic derivatives useful as anticancer agents
WO2000047212A1 (en) * 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
JP2000309577A (en) * 1999-01-27 2000-11-07 Pfizer Prod Inc Heteroaromatic bicyclic derivative useful as anticancer agent
JP2000514806A (en) * 1996-07-13 2000-11-07 グラクソ、グループ、リミテッド Fused heterocyclic compounds as protein tyrosine kinase inhibitors
WO2003040108A1 (en) * 2001-11-03 2003-05-15 Astrazeneca Ab Quinazoline derivatives as antitumor agents
WO2003040109A2 (en) * 2001-11-03 2003-05-15 Astrazeneca Ab Quinazoline derivatives as antitumor agents

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4642347A (en) * 1985-05-21 1987-02-10 American Home Products Corporation 3(2-quinolinylalkoxy)phenols
US5245038A (en) * 1987-11-06 1993-09-14 Baxter Diagnostics Inc. Fluorescent poly(arylpyridine) rare earth chelates
IL89029A (en) * 1988-01-29 1993-01-31 Lilly Co Eli Fungicidal quinoline and cinnoline derivatives, compositions containing them, and fungicidal methods of using them
US4952567A (en) * 1988-05-09 1990-08-28 City Of Hope Inhibition of lipogenesis
MX9200299A (en) * 1991-02-07 1992-12-01 Roussel Uclaf NEW NITROGENATED BICYCLE DERIVATIVES, THEIR PROCEDURE FOR PREPARING THE NEW INTERMEDIATE COMPOUNDS OBTAINED THEIR APPLICATION AS MEDICINES AND THE PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM.
EP0498722B1 (en) * 1991-02-07 1997-07-30 Roussel Uclaf Bicyclic nitrogen compounds, their preparation, intermediates obtained, their use as pharmaceuticals and pharmaceutical compositions containing them
US6004979A (en) * 1991-02-07 1999-12-21 Hoechst Marion Roussel Nitrogenous bicycles
US6645969B1 (en) * 1991-05-10 2003-11-11 Aventis Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US5245036A (en) * 1992-05-07 1993-09-14 Dowelanco Process for the preparation of 4-phenoxyquinoline compounds
US5972598A (en) * 1992-09-17 1999-10-26 Board Of Trustess Of The University Of Illinois Methods for preventing multidrug resistance in cancer cells
EP1195372A1 (en) * 1994-04-18 2002-04-10 Mitsubishi Pharma Corporation N-heterocyclic substituted benzamide derivatives with antihypertensive activity
US5840695A (en) * 1994-10-07 1998-11-24 Heska Corporation Ectoparasite saliva proteins and apparatus to collect such proteins
US5851545A (en) * 1995-08-25 1998-12-22 Sandoz Ltd. Insecticidal matrix and process for preparation thereof
US5906819A (en) * 1995-11-20 1999-05-25 Kirin Beer Kabushiki Kaisha Rho target protein Rho-kinase
DE19608653A1 (en) * 1996-03-06 1997-09-11 Thomae Gmbh Dr K Pyrimido [5,4-d] pyrimidines, medicaments containing these compounds, their use and processes for their preparation
JP3669711B2 (en) * 1996-08-12 2005-07-13 三菱ウェルファーマ株式会社 Medicament containing Rho kinase inhibitor
AR012634A1 (en) * 1997-05-02 2000-11-08 Sugen Inc QUINAZOLINE BASED COMPOUND, FAMACEUTICAL COMPOSITION THAT UNDERSTANDS IT, METHOD TO SYNTHESIZE IT, ITS USE, METHODS OF MODULATION OF THE DESERINE / TREONIN PROTEIN-KINASE FUNCTION AND IN VITRO METHOD TO IDENTIFY COMPOUNDS THAT MODULATE
US5885803A (en) * 1997-06-19 1999-03-23 Incyte Pharmaceuticals, Inc. Disease associated protein kinases
HUP0103386A3 (en) * 1998-08-21 2002-07-29 Parker Hughes Inst St Paul Use of quinazoline derivatives for producing pharmaceutical compositions having jak 3-inhibitor effect
US6184226B1 (en) * 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
AU2096100A (en) * 1998-12-23 2000-07-31 Bayer Aktiengesellschaft Polycarbonates with a low yellowness index
US6080747A (en) * 1999-03-05 2000-06-27 Hughes Institute JAK-3 inhibitors for treating allergic disorders
UA73993C2 (en) * 2000-06-06 2005-10-17 Астразенека Аб Quinazoline derivatives for the treatment of tumours and a pharmaceutical composition
MXPA03008658A (en) * 2001-03-23 2005-04-11 Bayer Ag Rho-kinase inhibitors.
GB0128108D0 (en) * 2001-11-23 2002-01-16 Astrazeneca Ab Therapeutic use
JP4505228B2 (en) * 2002-01-10 2010-07-21 バイエル・シェーリング・ファルマ・アクチェンゲゼルシャフト Rho-kinase inhibitor
WO2003062227A1 (en) * 2002-01-23 2003-07-31 Bayer Pharmaceuticals Corporation Rho-kinase inhibitors
DE602004014347D1 (en) * 2003-03-12 2008-07-24 Millennium Pharm Inc CHINAZOLIN DERIVATIVES AS TGF BETA INHIBITORS

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06336481A (en) * 1992-12-10 1994-12-06 Zeneca Ltd Quinazoline derivative, its preparation and medicinal composition having anticancerous action and containing same
WO1995015758A1 (en) * 1993-12-10 1995-06-15 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit csf-1r receptor tyrosine kinase
WO1996039145A1 (en) * 1995-06-06 1996-12-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Protein tyrosine kinase aryl and heteroaryl quinazoline compounds having selective inhibition of her-2 autophosphorylation properties
JPH11508906A (en) * 1995-07-13 1999-08-03 グラクソ、グループ、リミテッド Heterocyclic compounds and pharmaceutical compositions containing them
JP2000514806A (en) * 1996-07-13 2000-11-07 グラクソ、グループ、リミテッド Fused heterocyclic compounds as protein tyrosine kinase inhibitors
WO1999006396A1 (en) * 1997-07-29 1999-02-11 Warner-Lambert Company Irreversible bicyclic inhibitors of tyrosine kinases
WO1999035132A1 (en) * 1998-01-12 1999-07-15 Glaxo Group Limited Heterocyclic compounds
WO1999035146A1 (en) * 1998-01-12 1999-07-15 Glaxo Group Limited Bicyclic heteroaromatic compounds as protein tyrosine kinase inhibitors
GB2345486A (en) * 1999-01-11 2000-07-12 Glaxo Group Ltd Heteroaromatic protein tyrosine kinase inhibitors
WO2000044728A1 (en) * 1999-01-27 2000-08-03 Pfizer Products Inc. Substituted bicyclic derivatives useful as anticancer agents
JP2000309577A (en) * 1999-01-27 2000-11-07 Pfizer Prod Inc Heteroaromatic bicyclic derivative useful as anticancer agent
WO2000047212A1 (en) * 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2003040108A1 (en) * 2001-11-03 2003-05-15 Astrazeneca Ab Quinazoline derivatives as antitumor agents
WO2003040109A2 (en) * 2001-11-03 2003-05-15 Astrazeneca Ab Quinazoline derivatives as antitumor agents

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008534518A (en) * 2005-03-25 2008-08-28 サーフェイス ロジックス,インコーポレイティド Improved pharmacokinetic compound
WO2007142323A1 (en) * 2006-06-08 2007-12-13 Ube Industries, Ltd. Novel indazole derivative having spiro ring structure in side chain
JP2012229240A (en) * 2006-12-08 2012-11-22 Irm Llc Compound and composition as protein kinase inhibitors
WO2017150174A1 (en) * 2016-03-04 2017-09-08 株式会社シンスター・ジャパン Pharmaceutical composition containing 2,4-diaminoquinazoline derivative or salt thereof as active ingredient, and 2,4-diaminoquinazoline derivative having specific structure
JPWO2017150174A1 (en) * 2016-03-04 2019-06-13 株式会社 シンスター・ジャパン Pharmaceutical composition containing 2,4-diaminoquinazoline derivative or a salt thereof as an active ingredient, and 2,4-diaminoquinazoline derivative having a specific structure

Also Published As

Publication number Publication date
JP4320705B2 (en) 2009-08-26
DE60218138D1 (en) 2007-03-29
DE60218138T2 (en) 2007-09-20
ES2280517T3 (en) 2007-09-16
PT1370552E (en) 2007-04-30
UY27225A1 (en) 2002-10-31
MY142915A (en) 2011-01-31
EP1370552B1 (en) 2007-02-14
CY1108041T1 (en) 2013-09-04
US20100249159A1 (en) 2010-09-30
WO2002076977A2 (en) 2002-10-03
EP1370552A2 (en) 2003-12-17
US20030087919A1 (en) 2003-05-08
MXPA03008659A (en) 2005-04-08
TWI336328B (en) 2011-01-21
CA2441501A1 (en) 2002-10-03
DK1370552T3 (en) 2007-05-07
CA2441501C (en) 2010-09-14
AU2002245709A1 (en) 2002-10-08
WO2002076977A3 (en) 2002-12-12
ATE353889T1 (en) 2007-03-15
US20060142314A1 (en) 2006-06-29
HK1061029A1 (en) 2004-09-03
AR035792A1 (en) 2004-07-14
PE20020958A1 (en) 2002-11-14

Similar Documents

Publication Publication Date Title
JP4320705B2 (en) Rho kinase inhibitor
JP4329003B2 (en) Rho kinase inhibitor
JP4505228B2 (en) Rho-kinase inhibitor
CN102803227B (en) morpholinyl quinazoline
ES2427166T3 (en) Novel tricyclic heterocyclic compound
US7371765B2 (en) Quinoline derivatives having VEGF inhibiting activity
US20120238540A1 (en) Amino-pyrimidine compounds as inhibitors of ikk epsilon and/or tbk1
US6930114B2 (en) Pyrrolo (2.1a)dihydroisoquinolines and their use as phosphodiesterase 10a inhibitors
TW201144310A (en) [5,6] heterocyclic compound
KR101921764B1 (en) Pyrazolo-quinolines
CN105008353A (en) 2-acylaminothiazole derivative and salt thereof
KR20140097391A (en) Morpholinyl benzotriazine for use in cancer therapy
JP2004505965A (en) Compound
WO2022206795A1 (en) A cyclin-dependent kinase inhibitor
US20150336949A1 (en) 3-(Pyrazolyl)-1H-Pyrrolo[2,3-b]Pyridine Derivatives as Kinase Inhibitors

Legal Events

Date Code Title Description
A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20050322

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20081028

A601 Written request for extension of time

Free format text: JAPANESE INTERMEDIATE CODE: A601

Effective date: 20090127

A602 Written permission of extension of time

Free format text: JAPANESE INTERMEDIATE CODE: A602

Effective date: 20090203

A601 Written request for extension of time

Free format text: JAPANESE INTERMEDIATE CODE: A601

Effective date: 20090224

A602 Written permission of extension of time

Free format text: JAPANESE INTERMEDIATE CODE: A602

Effective date: 20090303

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20090309

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20090407

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20090416

TRDD Decision of grant or rejection written
A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20090519

A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20090521

R150 Certificate of patent or registration of utility model

Free format text: JAPANESE INTERMEDIATE CODE: R150

FPAY Renewal fee payment (event date is renewal date of database)

Free format text: PAYMENT UNTIL: 20120612

Year of fee payment: 3

FPAY Renewal fee payment (event date is renewal date of database)

Free format text: PAYMENT UNTIL: 20120612

Year of fee payment: 3

FPAY Renewal fee payment (event date is renewal date of database)

Free format text: PAYMENT UNTIL: 20130612

Year of fee payment: 4

LAPS Cancellation because of no payment of annual fees